WO2020232190A1 - Inhibiteurs de protéine kinase et leurs utilisations pour le traitement de maladies et de problèmes de santé - Google Patents

Inhibiteurs de protéine kinase et leurs utilisations pour le traitement de maladies et de problèmes de santé Download PDF

Info

Publication number
WO2020232190A1
WO2020232190A1 PCT/US2020/032784 US2020032784W WO2020232190A1 WO 2020232190 A1 WO2020232190 A1 WO 2020232190A1 US 2020032784 W US2020032784 W US 2020032784W WO 2020232190 A1 WO2020232190 A1 WO 2020232190A1
Authority
WO
WIPO (PCT)
Prior art keywords
nmr
mmol
mhz
cdcl
ripk3
Prior art date
Application number
PCT/US2020/032784
Other languages
English (en)
Inventor
Gregory Cuny
Alexei Degterev
Sameer NIKHAR
Siddharth Balachandran
Original Assignee
University Of Houston System
Trustees Of Tufts College
Institute For Cancer Research D/B/A The Research Intitute Of Fox Chase Cancer Center
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University Of Houston System, Trustees Of Tufts College, Institute For Cancer Research D/B/A The Research Intitute Of Fox Chase Cancer Center filed Critical University Of Houston System
Priority to CA3137869A priority Critical patent/CA3137869A1/fr
Priority to EP20729566.8A priority patent/EP3968991A1/fr
Priority to JP2021568650A priority patent/JP2022533182A/ja
Priority to AU2020275304A priority patent/AU2020275304A1/en
Priority to US17/595,406 priority patent/US20220194937A1/en
Publication of WO2020232190A1 publication Critical patent/WO2020232190A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses

Definitions

  • Protein kinases are important enzymes in cellular signal transduction. In many pathological conditions aberrant signal transduction occurs. Therefore, protein kinase inhibitors can be used as therapeutic agents for the treatment of various diseases.
  • Influenza A virus (IAV) infections account for up to 700,000 hospitalizations and 50,000 annual deaths in the US alone. Worryingly, even though highly pathogenic H5 and H7 strains of avian IAV are thus far limited in their spread between humans, they require only a small number of mutations to become transmissible.
  • the lung epithelium is a primary site of mammalian Influenza A virus pathology.
  • Influenza A virus is a negative- sense RNA vims of the family Orthomyxoviridae .
  • vims replication occurs within the gastrointestinal tract and is typically asymptomatic.
  • mammalian IAV strains replicate in the respiratory tract and produce symptoms ranging from mild cases of‘the flu’ to severe, sometimes lethal disease.
  • IAV infects epithelial cells along the entire respiratory tract. Transmission is associated with infection of the upper tract, whereas severe disease is associated with infection in the lower lung, with the extent of lung involvement correlating with disease outcomes in humans and animal models.
  • IAV As a lytic vims, IAV kills most lung cell types in which it replicates. But not all cell death by IAV is pathogenic. While programmed cell death is essential for early control of IAV replication and prevention of virus spread to the whole lung, the death of the lung epithelial layer is also one of the primary drivers of IAV infection-associated morbidity and mortalitylO. In particular, the loss of Type I airway epithelial cells (AECs, which are essential for gas exchange) above a threshold of -10% is strongly correlated with mortality in the mouse model of IAV infection. When cell death is well-controlled and apoptotic, it represents a host defense mechanism that limits both virus spread and immunopathology.
  • AECs Type I airway epithelial cells
  • Receptor-interacting protein kinase 3 (RIPK3) is a multi-functional protein involved in cell death pathways in various tissues.
  • RIPK3 kinase activity provides a protection in a variety of animal models of inflammatory and degenerative conditions, including sepsis, brain trauma, acute kidney injuries, lung injury associated with influenza (IAV) infection, atherosclerosis, and many others.
  • IAV influenza
  • RIPK3 was recently uncovered as a targetable signaling pathway that accounts for almost all IAV-activated pathogenic death in infected cells. This pathway is initiated when the host sensor protein DAI detects IAV genomic RNA and activates RIPK3 kinase.
  • necroptosis or programmed necrosis
  • RIPK3 also activates a parallel pathway of non-pathogenic cell death (apoptosis) that mediates vims clearance.
  • apoptosis non-pathogenic cell death
  • inhibitors of RIPK3 kinase function will be expected to ameliorate necrotic lung injury without affecting vims clearance, and potentially represent an entirely new strategy for treatment of IAV disease.
  • two separate studies showed that animals deficient in Ripk3 gene show resistance in the development of atherosclerosis due to the inhibition of macrophage cell death and systemic inflammation.
  • the present disclosure relates generally to compounds that demonstrate inhibitory activity of receptor interacting kinase 2 (RIPK2) inhibitory activity and/or Activin- like kinase 2 (ALK2) and/or receptor interacting kinase 3 (RIPK3).
  • RIPK2 receptor interacting kinase 2
  • ALK2 Activin- like kinase 2
  • RIPK3 receptor interacting kinase 3
  • RIPK2 mediates pro-inflammatory signaling and is an emerging therapeutic target in autoimmune and inflammatory diseases, such as inflammatory bowel disease (IBD) and multiple sclerosis.
  • RIPK2 inhibitors could provide therapeutic benefit in the treatment of these and other conditions.
  • Activin-like kinase 2 (ALK2) has been implicated in a number of diseases, such as bone disease (e.g. fibrodysplasia ossificans progressiva, ankylosing spondylitis), cardiovascular diseases (e.g. atherosclerosis and vascular calcification), some cancers (e.g. diffuse intrinsic pontine gliomas) and bums.
  • ALK2 Activin-like kinase 2
  • Many of these maladies also have an inflammatory component that could exacerbate the condition and/or worsen the clinical outcome.
  • Compounds that are either dual RIPK2/ALK2 inhibitors or that preferentially inhibit RIPK2 or ALK2 could provide therapeutic benefit in the treatment of these and other conditions.
  • the RIPK3 inhibitors potently and specifically inhibit RIPK3-driven necroptosis. These molecules are based on a pyrido[2,3-d]pyrimidine scaffold and target the ATP- and allosteric Glu-out pockets of RIPK3. This series is already ⁇ 10-fold more potent than previously reported RIPK3 inhibitor compounds at inhibiting necroptosis in cells. Importantly, compounds are identified that block necroptosis but do so without toxicity.
  • This disclosure describes a targetable signaling pathway that accounts for almost all IAV-activated pathogenic death in infected cells.
  • This pathway is initiated when the host sensor protein DAI detects IAV genomic RNA and activates RIPK3 kinase.
  • RIPK3 then triggers a form of cell death called necroptosis (or programmed necrosis), responsible for much of the lung injury seen during IAV infection. Fascinatingly, eliminating necroptosis not only drastically reduces lung damage and improves animal survival, but does so without impeding vims clearance. This is because RIPK3 also activates a parallel pathway of non-pathogenic cell death (apoptosis) that mediates vims clearance.
  • inhibitors of RIPK3 kinase function will ameliorate necrotic lung injury without affecting virus clearance, and represent an entirely new strategy for treatment of IAV disease.
  • RIPK3 is the central mediator of IAV-triggered epithelial cell death, or necroptosis. Necroptosis is activated by virus and microbial infections (including IAV), several innate-immune signaling pathways (most notably TNF, interferons, TLRs), certain pro- inflammatory stimuli (e.g., asbestos, oxidized LDL), and genotoxic stress. For decades, it was believed that the cell death accompanying acute infection by IAV was unspecified apoptosis, autophagy, or simply the passive, unprogrammed consequence of infection by a‘lytic’ virus.
  • the necroptosis arm requires RIPK3 kinase activity and targets pseudokinase mixed lineage kinase domain-like (MLKL).
  • the apoptosis arm requires FAS- associated protein with death domain (FADD) and caspase-8, and proceeds without need for the kinase activity of RIPK3. From within the necrosome, RIPK3 phosphorylates and activates MLKL, which then oligomerizes and traffics to the plasma membrane where it punches holes in the membrane, triggering cell swelling and lysis. Such necroptotic lysis of the cell causes the release of‘danger-associated molecular patterns’ (DAMPs) into the extracellular space, and is considered highly inflammatory.
  • DAMPs ‘danger-associated molecular patterns’
  • RIPK3 can also activate a parallel pathway of apoptosis via caspase-8, which promotes viral clearance. Importantly, this pathway of apoptosis does not require the catalytic activity of RIPK3, and can actually be facilitated by RIPK3 kinase blockade.
  • FIG. 1 shows RIPK3-mediated cell death or nectroptosis pathways, including those activated by tumor necrosis factor (TNF) (left) and IAV (right).
  • TNF tumor necrosis factor
  • IAV IAV
  • RIPK3 kinase blockade can prevent inflammatory pathology without affecting virus clearance.
  • Mice deficient in the DAI-RIPK3 cell death pathway cannot control IAV spread in the lung and succumb to this vims, highlighting the importance of DAI-RIPK3 signaling in anti-IAV host defense.
  • the necrotic branch downstream of RIPK3 is not only redundant for viral clearance, but actively promotes epithelial degradation and inflammation ⁇
  • selectively blocking necroptosis downstream of RIPK3 will prevent pathology without affecting beneficial virus clearance, which will still proceed normally via caspase-8-mediated apoptosis.
  • RIPK3 -mediated necroptosis requires the kinase activity of RIPK3, and is therefore pharmacologically readily targetable by the use of RIPK3 kinase inhibitors.
  • a clinically viable RIPK3 small-molecule inhibitor will represent a promising new therapeutic avenue for IAV-triggered necrotic death, epithelial degradation, inflammation, and consequent lung injury as seen in FIG. 1, right side.
  • RIPK3-mediated necroptosis potently amplifies inflammation in a variety of chronic conditions, including atherosclerosis and many TNF-associated pathologies, and underlies lung damage during acute IAV infection and ARDS. Moreover, RIPK3 can directly activate inflammatory gene expression independent of cell death, highlighting its centrality to host inflammatory processes. In agreement, genetic ablation of RIPK3 in mice ameliorated development of atherosclerotic plaques, almost-completely abolished TNF-triggered inflammatory shock, strongly reduced injury arising from chronically produced TNF, and eliminated IAV-induced necrotic lung damage without affecting vims clearance.
  • RIPK3 has been implicated in lipid and lysosomal storage diseases, such as Neiman-Pick and Gaucher disease, respectively. RIPK3 also has a pathogenic role in auto-immune diseases, including multiple sclerosis and lupus. Activity of RIPK3 has also been implicated in neurogeneration in different tissues and disease paradigms, indicating it to be a target for a broad range of neurodegenerative diseases. RIPK3 has also been found as a target in a broad range of ischemia-reperfusion injuries, such as stroke, myocardial infarction, kidney, retinal and liver ischemia and others
  • the present disclosure relates to a new panel of RIPK3 inhibitors, which may be referred to as the UH15 series.
  • UH15 series These compounds are based on a pyrido[2,3- dlpyrimidine scaffold, structurally distinct from previous RIPK3 inhibitor molecules. Members of this series are ⁇ 10-fold more potent than other disclosed RIPK3 inhibitors at blocking necroptosis.
  • the present UH15 compounds block RIPK3 without triggering apoptotic activity, and thus without the toxicity of other RIPK3 inhibitors. For most indications (e.g., TNF pathologies), blocking RIPK3 necroptosis without concurrently activating apoptosis is ideal.
  • a RIPK3 inhibitor that can efficiently block necroptosis while concurrently potentiating apoptosis of infected cells will not only halt deleterious necrotic lung damage but will also accelerate eradication of vims.
  • induction of apoptosis must be limited to the infected cell (i.e., be stimulus-specific) or toxicity will ensue (as seen with the previous compounds).
  • the present RIPK3 inhibitors target RIPK3 necroptosis as a new therapeutic entry-point for IAV disease.
  • Seasonal and pandemic strains of IAV trigger necrotic lung damage that underlies both ARDS and viral/bacterial pneumonia, each of which remain major causes of morbidity and mortality, with few effective therapeutic options.
  • the ideal therapeutic for severe IAV disease is one that blocks RIPK3 -dependent necroptosis but does not impede (or perhaps even simultaneously promotes) non-inflammatory virus clearance via RIPK3-dependent apoptosis.
  • RIPK3 is central to multiple inflammatory diseases, besides those seen during IAV infection. These include both acute and chronic inflammatory conditions known to be driven by TNF, such as colitis and rheumatoid arthritis. Moreover, RIPK3 is essential for all known pathways of necroptosis, while the allied kinase RIPK1 (currently the focus of efforts by others) only participates in a subset of necroptotic pathways, such as those activated by TNF (see FIG. 1, right). RIPK3 inhibitors thus will have benefit in a wider range of acute and chronic inflammatory conditions than serve as anti-TNF approaches or RIPK1 inhibitors.
  • FIG. 1 shows diagrams of RIPK3 -mediated cell death pathways activated by IAV (left) and TNF (right).
  • FIG. 2 shows structures of exemplary protein kinase inhibitors, identified as UH15 analogs.
  • FIG. 3 shows structures of exemplary RIPK3 inhibitors.
  • FIG. 4 shows a general overall synthetic scheme for exemplary preferred compounds disclosed herein as inhibitors of protein kinase activity, referred to generally as UH15 analogs, or UH15s.
  • FIG. 5 shows steps in the synthesis of an exemplary RIPK3 inhibitor.
  • FIG. 6 shows steps in the synthesis of an exemplary RIPK3 inhibitor.
  • FIG. 7 shows steps in the synthesis of an exemplary RIPK3 inhibitor.
  • FIGs. 8-9 show synthetic schemes for intermediate compounds used in the synthesis of exemplary inhibitors of RIPK3.
  • FIG. 10 shows structures for intermediate compounds used in the synthesis of exemplary inhibitors of RIPK3.
  • FIG. 11 shows a synthetic scheme for intermediate compounds used in the synthesis of exemplary inhibitors of RIPK3.
  • FIGs. 12-13 show structures for intermediate compounds used in the synthesis of exemplary inhibitors of RIPK3.
  • FIGs. 14-15 show synthetic schemes for intermediate compounds used in the synthesis of exemplary inhibitors of RIPK3.
  • FIG. 16 shows structures for intermediate compounds used in the synthesis of exemplary inhibitors of RIPK3.
  • FIG. 17 shows a synthetic scheme for exemplary inhibitors of RIPK3, in accordance with preferred embodiments.
  • FIGs. 18-21 show structures for exemplary inhibitors of RIPK3, in accordance with preferred embodiments.
  • FIGs. 22-23 show synthetic schemes for exemplary inhibitors of RIPK3, in accordance with preferred embodiments.
  • FIG. 24 A shows a synthetic scheme for exemplary inhibitors of RIPK3, in accordance with preferred embodiments.
  • FIG. 24B shows structures of intermediate compounds used in the synthesis of exemplary inhibitors of RIPK3, in accordance with preferred embodiments.
  • FIG. 25 shows structures of exemplary inhibitors of RIPK3, in accordance with preferred embodiments.
  • FIGs. 26-30 shows synthetic schemes for exemplary inhibitors of RIPK3, in accordance with preferred embodiments.
  • FIG. 31 shows activity of select UH15 compounds and GSK’872 in blocking RIPK3 and MLKL phosphorylation in RAW264.7 macrophages.
  • the present disclosure relates to protein kinase inhibitors and uses thereof.
  • FIG. 1 The following figure depicts general structures of preferred embodiments of compounds that inhibit protein kinases, including receptor interacting kinase 2 (RIPK2), Activin-like kinase 2 (ALK2), and receptor interacting kinase 3 (RIPK3).
  • RIPK2 receptor interacting kinase 2
  • ALK2 Activin-like kinase 2
  • RIPK3 receptor interacting kinase 3
  • A can be N or CH.
  • R1 can be Me, , or
  • R2 can be Me, Et, Et-O-Me, or isobutyl.
  • R 3 can be where Et is ethyl.
  • FIG. 2 Additional preferred embodiments of compounds that inhibit RIPK2, ALK2, and RIPK3 are shown in FIG. 2.
  • ALK2, and RIPK3 are shown below.
  • R2 is Et (UH15-4), Et-O-Me (UH15-6), or isobutyl (UH15-10), and where X is F (UH15-18) or Me (UH15-20).
  • R can be H, or R can be a substituent on any one available position of the phenyl ring that is SO 2 Me,
  • E can be N, CH, or C-R, with R defined as above.
  • B and C can independently be N, CH, or C-Cl.
  • R 1 can be H, or R 1 can be C-Cl, C-F, C-OCH 3 , C-C(CH 3 ) 3 , or C-OH at any one available position of the ring.
  • a and D can independently be N or CH.
  • E can be N, CH, or C-R.
  • B and C can independently be N, CH, or C-Cl.
  • alkylhydroxyl including but not limited to 2-hydroxyethyl, alkylalkoxyl, including but not limited to 2-methoxyethyl, or alkylaryl, including but not limited to benzyl or phenethyl.
  • R can be H, where Me is
  • R 1 can be any alkyl group, including but not limited to methyl, ethyl, or propyl, or R 1 can be any aryl group, including but not limited to naphthyl, thienyl, indoyl, and the like.
  • R 3 can be H, or R 3 can be C-Cl, C-F, C-OCH 3 , C-C(CH 3 ) 3 , or C-OH at any one available position of the ring.
  • exemplary compounds that inhibit protein kinases including receptor interacting kinase 2 (RIPK2), Activin-like kinase 2 (ALK2), and receptor interacting kinase 3 (RIPK3), described herein may occur in different geometric and enantiomeric forms, and both pure forms and mixtures of these separate isomers are included in the scope of this invention, as well as any physiologically functional or pharmacologically acceptable salt derivatives or prodrugs thereof. Production of these alternate forms would be well within the capabilities of one skilled in the art.
  • the current invention also pertains to methods of prevention or therapy for inflammatory and degenerative conditions, including diseases involving protein kinase activity, such as Influenza A virus (IAV) infections and inflammatory conditions driven by TNF, including the step of administering a compound that inhibits protein kinase activity in accordance with preferred embodiments disclosed herein.
  • the methods of prevention or therapy for inflammatory or degenerative diseases involving protein kinase activity include the step of administering a compound that is a compound shown in FIG. 2 or in FIG. 3.
  • a pharmaceutical composition including a therapeutically effective amount of a compound that inhibits protein kinase activity as defined above and a pharmaceutically acceptable excipient, adjuvant, carrier, buffer or stabiliser.
  • A“therapeutically effective amount” is to be understood as an amount of an exemplary protein kinase inhibitor compound that is sufficient to show inhibitory effects on protein kinase activity.
  • the actual amount, rate and time-course of administration will depend on the nature and severity of the disease being treated. Prescription of treatment is within the responsibility of general practitioners and other medical doctors.
  • the pharmaceutically acceptable excipient, adjuvant, carrier, buffer or stabiliser should be non-toxic and should not interfere with the efficacy of the active ingredient.
  • the precise nature of the carrier or other material will depend on the route of administration, which may be oral, or by injection, such as cutaneous, subcutaneous, or intravenous injection, or by dry powder inhaler.
  • compositions for oral administration may be in tablet, capsule, powder or liquid form.
  • a tablet may comprise a solid carrier or an adjuvant.
  • Liquid pharmaceutical compositions generally comprise a liquid carrier such as water, petroleum, animal or vegetable oils, mineral oil or synthetic oil. Physiological saline solution, dextrose or other saccharide solution or glycols such as ethylene glycol, propylene glycol or polyethylene glycol may be included.
  • a capsule may comprise a solid carrier such as gelatin.
  • the active ingredient will be in the form of a parenterally acceptable aqueous solution which is pyrogen-free and has a suitable pH, isotonicity and stability.
  • isotonic vehicles such as sodium chloride solution, Ringer’s solution, or lactated Ringer’s solution.
  • Preservatives, stabilisers, buffers, antioxidants and/or other additives may be included as required.
  • salt any acid or base derived salt formed from hydrochloric, sulfuric, phosphoric, acetic, citric, oxalic, malonic, salicylic, malic, fumaric, succinic, ascorbic, maleic, methanesulfonic, isoethonic acids and the like, and potassium carbonate, sodium or potassium hydroxide, ammonia, triethylamine, triethanolamine and the like.
  • prodrug means a pharmacological substance that is administered in an inactive, or significantly less active, form. Once administered, the prodrug is metabolised in vivo into an active metabolite.
  • the term“therapeutically effective amount” means a nontoxic but sufficient amount of the drug to provide the desired therapeutic effect.
  • the amount that is“effective” will vary from subject to subject, depending on the age and general condition of the individual, the particular concentration and composition being administered, and the like. Thus, it is not always possible to specify an exact effective amount. However, an appropriate effective amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation. Furthermore, the effective amount is the concentration that is within a range sufficient to permit ready application of the formulation so as to deliver an amount of the drug that is within a therapeutically effective range.
  • FIG. 4 shows a general overall synthetic scheme for exemplary preferred compounds disclosed herein as inhibitors of protein kinase activity, referred to generally as UH15 analogs, or UH15s.
  • UH15 analogs or UH15s.
  • compounds may be referred to in the format of, for example, UH15-1 or UH15_1. These compounds have different structures.
  • UH15-15 is a different compound from UH15_15.
  • NMR spectra were recorded at room temperature using a JEOL ECA- 500 (1H NMR and 13C NMR at 400, 500 and 600 MHz) with tetramethylsilane (TMS) as an internal standard. Chemical shifts (d) are given in parts per million (ppm) with reference to solvent signals [1H-NMR: CDCl 3 (7.26 ppm), CD 3 OD (3.30 ppm), DMSO-d 6 (2.49 ppm); 13C-NMR: CDCl 3 (77.0 ppm), CD 3 OD (49.0 ppm), DMSO-d 6 (39.5 ppm)].
  • A21 was synthesized by using the using adaptation of the methods described in Cuny, et al. WO 2018/213219.
  • reaction mixture was then partitioned between ethyl acetate and water, dried over anhydrous Na 2 SO 4 , filtered, concentrated and purified by column chromatography using silica gel (20 % EtOAc/DCM) to give Nap-5 (23 mg, 36%) as light yellow solid.
  • FIGs. 8-9, 11, and 14-15 show synthetic schemes for intermediate compounds used in the synthesis of exemplary inhibitors of protein kinase, in accordance with preferred embodiments.
  • FIGs. 10, 12-13, and 16 show structures for intermediate compounds used in the synthesis of exemplary inhibitors of protein kinase, in accordance with preferred embodiments.
  • Ethyl 4-amino-2-(methylthio)pyrimidine-5-carboxylate (2) [0108] To a solution of ethyl 4-chloro-2-(methylthio)pyrimidine-5-carboxylate (1) (100 mg, 0.43 mmol) in dry THF (2 mL) was added triethylamine (0.2 mL, 1.29 mmol) and ammonium hydroxide (0.5 mL). The resulting mixture was stirred at rt for 2 h till completion. After evaporation in vacuo to remove THF, the crude mixture then partitioned between H 2 O and EtOAc.
  • Method A Formic acid (1 mL) was added to 15a (100 mg, 0.48 mmol) in round bottom flask containing molecular sieves. The reaction mixture was heated at 60 °C for 6 h and then partitioned between saturated solution of NaHCO 3 and EtOAc. The organic layer was then washed with brine solution and then concentrated to give 16a (90 mg, 79%) as brown viscous liquid which was used directly into next step without purification.
  • 16c was prepared by method A where the reaction mixture was stirred at room temperature with overnight stirring. Yield 80%, white solid; 1 H NMR (400 MHz, DMSO- d6) d (ppm) 10.64 (s, 1H), 8.40 (s, 1H), 8.28 (s, 1H), 7.89-7.86 (m, 1H), 7.67-7.63 (m, 2H), 3.24 (s, 3H). 13 C NMR (100 MHz, DMSO-d 6 ) d 160.74, 141.95, 139.44, 130.80, 124.15, 122.51, 117.54, 44.08.
  • Method B To a round bottom flask fitted with reflux condenser was added 15d (200 mg, 0.72 mmol) and ethyl formate (1.16 mL, 14.4 mmol). TEA (0.15 mL, 1.08 mmol) was added to the above mixture and heated under reflux. After overnight stirring, solvent was evaporated, and the mixture was dissolved in DCM and extracted with water and brine solution. The extract was concentrated and purified by column chromatography using silica gel (30% EtOAc/Hexane) to get 16d (lOOmg, 45%) as colourless liquid.
  • Method C Formic acid (0.28 mL, 7.43 mmol) was added dropwise to acetic anhydride (0.60 mL, 6.37 mmol) maintained at 0 °C. The mixture was heated to reflux at 60 °C for 2 h to generate acetic formic anhydride reagent. The mixture was cooled to room temperature and 2 mL THF was added. 15e (200 mg, 2.12 mmol) dissolved in THF (1 mL) was added to acetic formic anhydride mixture and refluxed for another 2 h. Solvent was evaporated after completion, extracted in EtOAc/water system. Extract was concentrated and purified by column chromatography using silica gel (3% MeOH/DCM) to get 16e (173 mg, 67%) as white solid.
  • 16f was prepared by method C: Yield 75%, white solid.
  • FIG. 17 shows a synthetic scheme for exemplary inhibitors of RIPK3, compounds 17a-s (UH15’s).
  • FIGs. 18-21 show structures for exemplary inhibitors of RIPK3, in accordance with preferred embodiments.
  • 16a (30 mg, 0.13 mmol) and 14i (40 mg, 0.12 mmol) were used to make 17i.
  • 16a 22 mg, 0.09 mmol
  • 14k 20 mg, 0.05 mmol
  • 16a (16 mg, 0.07 mmol) and 141 (32 mg, 0.07 mmol) were used to make 171.
  • 16a 32 mg, 0.14 mmol
  • 14e 50 mg, 0,14 mmol
  • 16c (12mg, 0.06 mmol) and 14s (20 mg, 0.05 mmol) were used to make 17s.
  • 16a (8 mg, 0.03 mmol) and 14s (14 mg, 0.03 mmol) were used to make 17s * .
  • FIG. 22 shows a synthetic scheme for an exemplary inhibitor of protein kinase, compound UH15_32, in accordance with preferred embodiments.
  • Ethyl 6-chloro-4-(methylamino)nicotinate (52): [0324] To a solution of Ethyl 4,6-dichloronicotinate 51 (lOOmg, 0.45 mmol) in THF (3 mL) was added aqueous methyl amine (0.4 mL) at 0 °C and the mixture was stirred for 30 min at the same temperature. After 30 min the reaction mixture was stirred at room temperature for 2 h. Following completion the mixture was concentrated and purified by column chromatography using silica gel (10% EtO Ac/Hexane) to afford 52 (65 mg, 67%) as white solid.
  • FIG. 23 shows a synthetic scheme for an exemplary inhibitor of protein kinase, compound UH15_33, in accordance with preferred embodiments.
  • FIG. 24A shows a synthetic scheme for exemplary inhibitors of protein kinases and FIG. 24B shows the structures of intermediate compounds used in the synthesis of these exemplary inhibitors of protein kinases, in accordance with preferred embodiments.
  • FIG. 25 shows structures of exemplary inhibitors of protein kinases, in accordance with preferred embodiments.
  • FIG. 26 shows a synthetic scheme for exemplary inhibitors of protein kinase UH15PN 5 , in accordance with preferred embodiments.
  • FIG. 27 shows a synthetic scheme for an exemplary inhibitor of protein kinase UH15_25, in accordance with preferred embodiments.
  • reaction mixture was then partitioned between ethyl acetate and water, dried over anhydrous Na 2 SO 4 , filtered, concentrated and purified by column chromatography using silica gel (1.5 % MeOH/DCM) to give 60 (20 mg, 57%) as light yellow solid.
  • FIG. 28 shows a synthetic scheme for exemplary inhibitor of protein kinase UH15_36, in accordance with preferred embodiments.
  • reaction mixture was then partitioned between ethyl acetate and water, dried over anhydrous Na 2 SO 4 , filtered, concentrated and purified by column chromatography using silica gel (2.5 % MeOH/DCM) to give 71 (26 mg, 35%) as light brown solid.
  • FIG. 29 shows a synthetic scheme for exemplary inhibitor of protein kinase UH15_20, in accordance with preferred embodiments.
  • FIG. 30 shows a synthetic scheme for an exemplary inhibitor of protein kinase UH15_20, in accordance with preferred embodiments.
  • Reagents and conditions (a) TBSC1, imidazole, DMAP, DMF, 0 °C to rt, 4 h, 83%; (b) KF/AI 2 O 3 , DMA, rt, 2 h, 67%; (c) Pd2(dba)3, Xantphos, CS2CO 3 , 1,4-dioxane, 80 °C, 16 h, 47%.
  • Table 1 below shows the activities of select UH15 analogs.
  • IC 50 /LD50 values were determined for inhibition of recombinant RIPK3, inhibition of necroptosis in LPS/IDN- treated murine RAW264.7 macrophages and TNF/IDN-treated human Hela cells expressing RIPK3, RIPK3 binding by the inhibitors in live cells (NanoBRET assay), and induction of RIPK3 -dependent apoptosis (i.e., on-target toxicity).
  • Assays were performed using ADPGlo (kinase assay), CellTiter-Glo (viability), and NanoBRET (target engagement) assays.
  • the GSK’872 value shown is from Mandal et al. 2014.
  • recombinant RIPK3 protein (20 ng per reaction) is diluted in the reaction buffer consisting of 50 mM HEPES, pH 7.5, 50 mM NaCl, 30 mM MgC12, 1 mM DTT, 0.05% bovine serum albumin (BSA), 0.02% CHAPS.
  • Diluted protein is added to low volume white 384 well plates (2 mL/well).
  • Inhibitors are diluted in reaction buffer (final 25% DMSO), 1 mL is added to each well and incubated 5 min at room temperature. Reactions are initiated by the addition of 2 mL of 100 pM ATP in the reaction buffer.
  • IC 50 values are calculated based on a dose range of inhibitor concentrations using non-linear regression in GraphPad Prism software.
  • hTNFa Peprotech, Hela-R 3 cells
  • E.coli LPS E.coli LPS
  • 10 mM IDN6556 MedKoo
  • FIG. 31 shows that select UH15 compounds and GSK’ 872 block RIPK3 and MLKL phosphorylation in RAW264.7 macrophages.
  • Necroptosis was induced by the combination of LPS (10 ng/ml) and pan-caspase inhibitor IDN6556 (20 mM).
  • LPS is a TLR4 ligand that, like TNF, can activate RIPK3 (and necroptosis) in the presence of a caspase inhibitor.
  • the asterisk indicates a non-specific band sometimes see in pMLKL immunoblots. Samples are subjected to SDS-PAGE gele electrophoresis and Western blotting using RIPK3, phospho-Thr231/Ser232-RIPK3, MLKL and phospho-Ser345-MLKL antibodies (Abeam).
  • Table 2 shows stimulus-dependent (with TNF, 10 ng/ml) and -independent (without TNF) RIPK3 -dependent toxicity of select UH15 analogs.
  • Cell death was assessed by CellTiter-Glo assay. Toxicity was compared in Hela cells lacking RIPK3 and cells where RIPK3 was lentivirally re-expressed (Hela-RIPK3 cells).
  • RIPK2 Receptor interacting protein kinase 2
  • Recombinant RIPK2 protein (20 ng per reaction) is diluted in the reaction buffer consisting of 40 mM Tris (pH 7.5); 20 mM MgCl 2 ; 0.1 mg/mL BSA; 50 mM DTT. Diluted protein is added to low volume white 384 well plates (2 mL/well). Inhibitors are diluted in reaction buffer (final 25% DMSO), 1 mL is added to each well and incubated 5 min at room temperature. Reactions are initiated by the addition of 2 mL of 100 pM ATP and 1 mg/mL RS repeat peptide (SignalChem) in the reaction buffer. Plates are sealed with plastic coverslips and incubated at room temperature for 2 h.
  • ALK2 Activin-like kinase 2
  • Enzyme inhibitory activity was evaluated in a standard kinase enzyme assay by incubating human ALK2 with the protein substrate casein (1 mg/mL) and g- 33 ATR (10 pM) in the presence of various concentrations of test compounds (10 nM - 100 pM). After 30 min the amount of 33 P-casein was determined. A plot of inhibitor concentration verses % activity was constructed and from this plot an IC 50 value was determined.
  • HEK-Blue cells expressing human NOD2 and NFkB-SAEP reporter are seeded into 96 well clear plates at 7.5x10 3 cells per well in 100 mL of DMEM media supplemented with 10% FBS and 1% antibiotic-antimycotic mix. Cells are allowed to attach for 48 h in 5% CO 2 tissue culture incubator at 37 °C. On the morning of the experiment, media in the wells is replaced with 100 mL of HEK-Blue detection media (Invivogen). Cells are treated with the inhibitors, diluted in DMSO (0.5 mL per well) for 15 min in 5% CO 2 tissue culture incubator at 37 °C.
  • IC 50 values are calculated based on a dose range of inhibitor concentrations using non-linear regression in GraphPad Prism software.
  • Rodrigue-Gervais I.G. et al.
  • Cellular inhibitor of apoptosis protein cIAP2 protects against pulmonary tissue necrosis during influenza virus infection to promote host survival. Cell Host Microbe 15, 23-35 (2014).

Abstract

La présente invention concerne des composés identifiés démontrant une activité inhibitrice de la protéine kinase. Plus spécifiquement, les composés sont mis en évidence pour inhiber la kinase 2 interagissant avec le récepteur (RIPK2) et/ou la kinase 2 de type activine (ALK2) et/ou la kinase 3 interagissant avec le récepteur (RIPK3). Les composés qui sont des inhibiteurs doubles RIPK2/ALK2 ou qui inhibent de préférence RIPK2 ou ALK2 pourraient fournir un bénéfice thérapeutique. Les composés qui fonctionnent en tant qu'inhibiteurs de RIPK3 fournissent un bénéfice thérapeutique dans le traitement des problèmes inflammatoires et dégénératifs.
PCT/US2020/032784 2019-05-16 2020-05-14 Inhibiteurs de protéine kinase et leurs utilisations pour le traitement de maladies et de problèmes de santé WO2020232190A1 (fr)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CA3137869A CA3137869A1 (fr) 2019-05-16 2020-05-14 Inhibiteurs de proteine kinase et leurs utilisations pour le traitement de maladies et de problemes de sante
EP20729566.8A EP3968991A1 (fr) 2019-05-16 2020-05-14 Inhibiteurs de protéine kinase et leurs utilisations pour le traitement de maladies et de problèmes de santé
JP2021568650A JP2022533182A (ja) 2019-05-16 2020-05-14 タンパク質キナーゼ阻害剤ならびに疾患および状態の治療のためのその使用
AU2020275304A AU2020275304A1 (en) 2019-05-16 2020-05-14 Protein kinase inhibitors and uses thereof for the treatment of diseases and conditions
US17/595,406 US20220194937A1 (en) 2019-05-16 2020-05-14 Protein kinase inhibitors and uses thereof for the treatment of diseases and conditions

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201962848648P 2019-05-16 2019-05-16
US201962848719P 2019-05-16 2019-05-16
US62/848,719 2019-05-16
US62/848,648 2019-05-16

Publications (1)

Publication Number Publication Date
WO2020232190A1 true WO2020232190A1 (fr) 2020-11-19

Family

ID=70919255

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/032784 WO2020232190A1 (fr) 2019-05-16 2020-05-14 Inhibiteurs de protéine kinase et leurs utilisations pour le traitement de maladies et de problèmes de santé

Country Status (6)

Country Link
US (1) US20220194937A1 (fr)
EP (1) EP3968991A1 (fr)
JP (1) JP2022533182A (fr)
AU (1) AU2020275304A1 (fr)
CA (1) CA3137869A1 (fr)
WO (1) WO2020232190A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113072471A (zh) * 2021-03-02 2021-07-06 四川美大康华康药业有限公司 一种利非司特中间体及其制备方法

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018213219A1 (fr) * 2017-05-15 2018-11-22 University Of Houston System Pyrido[2,3-d]pyrimidin-7ones et composés apparentés utilisés en tant qu'inhibiteurs de protéine kinases

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996034867A1 (fr) * 1995-05-03 1996-11-07 Warner-Lambert Company PYRIDO[2,3-d]PYRIMIDINES DESTINEES A INHIBER LA PROLIFERATION CELLULAIRE INDUITE PAR LES TYROSINES KINASES
WO1999009030A1 (fr) * 1997-08-20 1999-02-25 Warner-Lambert Company Naphtyridinones utiles pour inhiber la proteine tyrosine kinase et une proliferation cellulaire dependant de kinases de cycle cellulaire
WO2001055148A1 (fr) * 2000-01-27 2001-08-02 Warner-Lambert Company Derives de pyridopyrimidinone destines au traitement de maladies maladie neurodegenerative
WO2014031571A1 (fr) * 2012-08-21 2014-02-27 Icahn School Of Medicine At Mount Sinai Traitement d'infections virales
WO2015179436A1 (fr) * 2014-05-19 2015-11-26 Sanford-Burnham Medical Research Institute Traitement de l'inflammation au moyen d'inhibiteurs de mekk3 ou de peptides bloquants
WO2018213219A1 (fr) 2017-05-15 2018-11-22 University Of Houston System Pyrido[2,3-d]pyrimidin-7ones et composés apparentés utilisés en tant qu'inhibiteurs de protéine kinases

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996034867A1 (fr) * 1995-05-03 1996-11-07 Warner-Lambert Company PYRIDO[2,3-d]PYRIMIDINES DESTINEES A INHIBER LA PROLIFERATION CELLULAIRE INDUITE PAR LES TYROSINES KINASES
WO1999009030A1 (fr) * 1997-08-20 1999-02-25 Warner-Lambert Company Naphtyridinones utiles pour inhiber la proteine tyrosine kinase et une proliferation cellulaire dependant de kinases de cycle cellulaire
WO2001055148A1 (fr) * 2000-01-27 2001-08-02 Warner-Lambert Company Derives de pyridopyrimidinone destines au traitement de maladies maladie neurodegenerative
WO2014031571A1 (fr) * 2012-08-21 2014-02-27 Icahn School Of Medicine At Mount Sinai Traitement d'infections virales
WO2015179436A1 (fr) * 2014-05-19 2015-11-26 Sanford-Burnham Medical Research Institute Traitement de l'inflammation au moyen d'inhibiteurs de mekk3 ou de peptides bloquants
WO2018213219A1 (fr) 2017-05-15 2018-11-22 University Of Houston System Pyrido[2,3-d]pyrimidin-7ones et composés apparentés utilisés en tant qu'inhibiteurs de protéine kinases

Non-Patent Citations (30)

* Cited by examiner, † Cited by third party
Title
AAES, T.L. ET AL.: "Vaccination with Necroptotic Cancer Cells Induces Efficient Anti-tumor Immunity", CELL REP, vol. 15, 2016, pages 274 - 287
ARRAZOLA, M.S. ET AL.: "Axonal degeneration is mediated by necroptosis activation", J NEUROSCI, 2019
ARRAZOLA, M.S.COURT, F.A.: "Compartmentalized necroptosis activation in excitotoxicity-induced axonal degeneration: a novel mechanism implicated in neurodegenerative disease pathology", NEURAL REGEN RES, vol. 14, 2019, pages 1385 - 1386
COUGNOUX, A. ET AL.: "Necroptosis in Niemann-Pick disease, type C1: a potential therapeutic target", CELL DEATH DIS, vol. 7, 2016, pages e2147
DUPREZ, L. ET AL.: "RIP kinase-dependent necrosis drives lethal systemic inflammatory response syndrome", IMMUNITY, vol. 35, 2011, pages 908 - 918, XP028348599, DOI: 10.1016/j.immuni.2011.09.020
KEARNEY, C.J.MARTIN, S.J.: "An Inflammatory Perspective on Necroptosis", MOL CELL, vol. 65, 2017, pages 965 - 973, XP029959277, DOI: 10.1016/j.molcel.2017.02.024
LI, J. ET AL.: "The RIP1/RIP3 necrosome forms a functional amyloid signaling complex required for programmed necrosis", CELL, vol. 150, 2012, pages 339 - 350, XP028930200, DOI: 10.1016/j.cell.2012.06.019
LIN, J. ET AL.: "A role of RIP3-mediated macrophage necrosis in atherosclerosis development", CELL REP, vol. 3, 2013, pages 200 - 210
LINKERMANN, A. ET AL.: "Necroptosis in immunity and ischemia-reperfusion injury", AM J TRANSPLANT, vol. 13, 2013, pages 2797 - 2804, XP055363009, DOI: 10.1111/ajt.12448
MANDAL, P. ET AL.: "RIP3 induces apoptosis independent of pronecrotic kinase activity", MOL CELL, vol. 56, 2014, pages 481 - 495, XP055363005, DOI: 10.1016/j.molcel.2014.10.021
MENG, L. ET AL.: "RIP3-dependent necrosis induced inflammation exacerbates atherosclerosis", BIOCHEM BIOPHYS RES COMMUN, vol. 473, 2016, pages 497 - 502, XP029510923, DOI: 10.1016/j.bbrc.2016.03.059
MENG, L.JIN, W.WANG, X.: "RIP3-mediated necrotic cell death accelerates systematic inflammation and mortality", PROC NATL ACAD SCI U S A, vol. 112, 2015, pages 11007 - 11012
MOQUIN, D.M.MCQUADE, T.CHAN, F.K.: "CYLD deubiquitinates RIP1 in the TNFalpha-induced necrosome to facilitate kinase activation and programmed necrosis", PLOS ONE, vol. 8, 2013, pages e76841
MORIWAKI, K.CHAN, F.K.: "Necrosis-dependent and independent signaling of the RIP kinases in inflammation", CYTOKINE GROWTH FACTOR REV, vol. 25, 2014, pages 167 - 174
MORIWAKI, K.CHAN, F.K.: "RIP3: a molecular switch for necrosis and inflammation", GENES DEV, vol. 27, 2013, pages 1640 - 1649
NAJJAR, M. ET AL.: "RIPK1 and RIPK3 Kinases Promote Cell-Death-Independent Inflammation by Toll-like Receptor 4", IMMUNITY, vol. 45, 2016, pages 46 - 59, XP029648398, DOI: 10.1016/j.immuni.2016.06.007
NEWTON, K. ET AL.: "RIPK3 deficiency or catalytically inactive RIPK1 provides greater benefit than MLKL deficiency in mouse models of inflammation and tissue injury", CELL DEATH DIFFER, vol. 23, 2016, pages 1565 - 1576
NOGUSA, S. ET AL.: "RIPK3 Activates Parallel Pathways of MLKL-Driven Necroptosis and FADD-Mediated Apoptosis to Protect against Influenza A Virus", CELL HOST MICROBE, vol. 20, 2016, pages 13 - 24, XP029639423, DOI: 10.1016/j.chom.2016.05.011
OFENGEIM, D. ET AL.: "Activation of necroptosis in multiple sclerosis", CELL REP, vol. 10, 2015, pages 1836 - 1849
PASPARAKIS, M.VANDENABEELE, P.: "Necroptosis and its role in inflammation", NATURE, vol. 517, 2015, pages 311 - 320
RODRIGUE-GERVAIS, I.G. ET AL.: "Cellular inhibitor of apoptosis protein cIAP2 protects against pulmonary tissue necrosis during influenza virus infection to promote host survival", CELL HOST MICROBE, vol. 15, 2014, pages 23 - 35, XP028810315, DOI: 10.1016/j.chom.2013.12.003
SALEH, D. ET AL.: "Kinase Activities of RIPK1 and RIPK3 Can Direct IFN-beta Synthesis Induced by Lipopolysaccharide", J IMMUNOL, 2017
SARHAN, J. ET AL.: "Constitutive interferon signaling maintains critical threshold of MLKL expression to license necroptosis", CELL DEATH DIFFER, vol. 26, 2019, pages 332 - 347, XP036721095, DOI: 10.1038/s41418-018-0122-7
SILKE, J.RICKARD, J.A.GERLIC, M.: "The diverse role of RIP kinases in necroptosis and inflammation", NAT IMMUNOL, vol. 16, 2015, pages 689 - 697
VANDEN BERGHE, T.HASSANNIA, B.VANDENABEELE, P.: "An outline of necrosome triggers", CELL MOL LIFE SCI, vol. 73, 2016, pages 2137 - 2152, XP035858591, DOI: 10.1007/s00018-016-2189-y
VITNER, E.B. ET AL.: "RIPK3 as a potential therapeutic target for Gaucher's disease", NAT MED, vol. 20, 2014, pages 204 - 208, XP055417593, DOI: 10.1038/nm.3449
VLANTIS, K. ET AL.: "NEMO Prevents RIP Kinase 1-Mediated Epithelial Cell Death and Chronic Intestinal Inflammation by NF-kappaB-Dependent and -Independent Functions", IMMUNITY, vol. 44, 2016, pages 553 - 567, XP029449039, DOI: 10.1016/j.immuni.2016.02.020
YUAN, J.AMIN, P.OFENGEIM, D.: "Necroptosis and RIPK1-mediated neuroinflammation in CNS diseases", NAT REV NEUROSCI, vol. 20, 2019, pages 19 - 33, XP036657643, DOI: 10.1038/s41583-018-0093-1
ZHANG, J.YANG, Y.HE, W.SUN, L.: "Necrosome core machinery: MLKL", CELL MOL LIFE SCI, vol. 73, 2016, pages 2153 - 2163, XP035858592, DOI: 10.1007/s00018-016-2190-5
ZHAO, H. ET AL.: "Role of necroptosis in the pathogenesis of solid organ injury", CELL DEATH DIS, vol. 6, 2015, pages e1975

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113072471A (zh) * 2021-03-02 2021-07-06 四川美大康华康药业有限公司 一种利非司特中间体及其制备方法

Also Published As

Publication number Publication date
US20220194937A1 (en) 2022-06-23
EP3968991A1 (fr) 2022-03-23
AU2020275304A1 (en) 2021-12-16
CA3137869A1 (fr) 2020-11-19
JP2022533182A (ja) 2022-07-21

Similar Documents

Publication Publication Date Title
KR102001745B1 (ko) 키나제 억제제로서 유용한 인돌 카르복스아미드 화합물
CN102124005B (zh) cMET抑制剂
TWI393566B (zh) 作為週期素依賴性激酶之新穎吡唑并嘧啶
US9567342B2 (en) Certain protein kinase inhibitors
US10087195B2 (en) Certain protein kinase inhibitors
KR102007056B1 (ko) 과증식성 질환 치료시 Bub1 키나제 저해제로 사용하기 위한 치환된 벤질인다졸
TW201906848A (zh) 化學化合物
US9024021B2 (en) Diarylacetylene hydrazide containing tyrosine kinase inhibitors
CA3142340A1 (fr) Inhibiteurs heterobicycliques de mat2a et procedes d'utilisation pour le traitement du cancer
EA021421B1 (ru) Производные хинолинов и хиноксалинов в качестве ингибиторов протеинтирозинкиназы, способ их получения, содержащая их фармацевтическая композиция и способ лечения заболеваний с применением таких соединений
USRE48974E1 (en) (5,6-dihydro)pyrimido[4,5-E]indolizines
Johns et al. Pyrazolopyridine antiherpetics: SAR of C2′ and C7 amine substituents
WO2014145642A9 (fr) Inhibiteurs de nrf2 à petite molécule pour traitement anticancéreux
KR20070027723A (ko) 4,6-이치환된 피리미딘 및 단백질 키나제 억제제로서의이의 용도
JP2017039765A (ja) タンパク質キナーゼ阻害薬
WO2014079232A1 (fr) Dérivés de 7-oxo-pyrimidine, compositions pharmaceutiques et utilisations de celles-ci
US20220194937A1 (en) Protein kinase inhibitors and uses thereof for the treatment of diseases and conditions
AU2002247059B2 (en) Method of treating inflammatory and immune diseases using inhibitors of IkappaB kinase (IKK)
US20220064163A1 (en) PYRIDO[2,3-d]PYRIMIDIN-7-ONES AND RELATED COMPOUNDS AS INHIBITORS OF PROTEIN KINASES
AU2004283093A1 (en) Compounds and compositions as protein kinase inhibitors
AU2015317937B2 (en) Preparation method for aromatic heterocyclic compound used as selective JAK3 and/or JAK1 kinase inhibitor and application of aromatic heterocyclic compound
JP2023512038A (ja) 化合物及びその使用
CA3214900A1 (fr) Polytherapies comprenant des inhibiteurs de mytl
CN103570731B (zh) 嘧啶并三环或嘧啶并四环类化合物及其药用组合物和应用
KR102269630B1 (ko) 2-아릴카보닐하이드라진카보티오아마이드 유도체 화합물 또는 이의 약학적으로 허용가능한 염을 포함하는 중동호흡기증후군 코로나바이러스 감염 질환의 예방 또는 치료용 약학적 조성물

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 20729566

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3137869

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021568650

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2020275304

Country of ref document: AU

Date of ref document: 20200514

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2020729566

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2020729566

Country of ref document: EP

Effective date: 20211216